Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 339
Filtrar
1.
Food Chem Toxicol ; 183: 114305, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38052405

RESUMO

Triclosan (TCS) is an antimicrobial compound incorporated into more than 2000 consumer products. This compound is frequently detected in the human body and causes ubiquitous contamination in the environment, thereby raising concerns about its impact on human health and environmental pollution. Here, we demonstrated that 20 weeks' exposure of TCS drove the development of glucose intolerance by inducing compositional and functional alterations in intestinal microbiota in rats. Fecal-transplantation experiments corroborated the involvement of gut microbiota in TCS-induced glucose-tolerance impairment. 16S rRNA gene-sequencing analysis of cecal contents showed that TCS disrupted the gut microbiota composition in rats and increased the ratio of Firmicutes to Bacteroidetes. Cecal metabolomic analyses detected that TCS altered host metabolic pathways that are linked to host glucose and amino acid metabolism, particularly branched-chain amino acid (BCAA) biosynthesis. BCAA measurement confirmed the increase in serum BCAAs in rats exposed to TCS. Western blot and immunostaining results further confirmed that elevated BCAAs stimulated mTOR, a nutrient-sensing complex, and following IRS-1 serine phosphorylation, resulted in insulin resistance and glucose intolerance. These results suggested that TCS may induce glucose metabolism imbalance by regulating BCAA concentration by remodeling the gut microbiota.


Assuntos
Microbioma Gastrointestinal , Intolerância à Glucose , Triclosan , Humanos , Ratos , Animais , Intolerância à Glucose/induzido quimicamente , Triclosan/toxicidade , RNA Ribossômico 16S/genética , Glucose
2.
J Endocrinol ; 260(1)2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37855335

RESUMO

Reports in North America suggest that up to 20% of young women (18-24 years) use cannabis during pregnancy. This is concerning given clinical studies indicate that maternal cannabis use is associated with fetal growth restriction and dysglycemia in the offspring. Preclinical studies demonstrated that prenatal exposure to Δ9-tetrahydrocannabinol, the main psychoactive component of cannabis, in rat dams led to female-specific deficits in ß-cell mass and glucose intolerance/insulin resistance. Yet to date, the contributions of cannabidiol (CBD), the primary nonpsychoactive compound in cannabis, remain elusive. This study aimed to define the effects of in utero cannabidiol (CBD) exposure on postnatal glucose regulation. Pregnant Wistar rat dams received daily intraperitoneal injections of either a vehicle solution or 3 mg/kg of CBD from gestational day (GD) 6 to parturition. CBD exposure did not lead to observable changes in maternal or neonatal outcomes; however, by 3 months of age male CBD-exposed offspring exhibited glucose intolerance despite no changes in pancreatic ß/α-cell mass. Transcriptomic analysis on the livers of these CBD-exposed males revealed altered gene expression of circadian rhythm clock machinery, which is linked to systemic glucose intolerance. Furthermore, alterations in hepatic developmental and metabolic processes were also observed, suggesting gestational CBD exposure has a long-lasting detrimental effect on liver health throughout life. Collectively, these results indicate that exposure to CBD alone in pregnancy may be detrimental to the metabolic health of the offspring later in life.


Assuntos
Canabidiol , Intolerância à Glucose , Resistência à Insulina , Células Secretoras de Insulina , Gravidez , Ratos , Feminino , Masculino , Humanos , Animais , Lactente , Canabidiol/toxicidade , Intolerância à Glucose/induzido quimicamente , Ratos Wistar
3.
J Trace Elem Med Biol ; 80: 127299, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37690370

RESUMO

BACKGROUND: Epidemiological studies have shown that exposure to sodium arsenite (NaAsO2) causes diabetes and hepatotoxicity. Metformin (MET), an oral hypoglycemic agent, has long been used in diabetes therapy. In addition, MET has been shown to have hepatoprotective effects. In this study, we investigated the effects of MET on NaAsO2-induced hepatotoxicity and glucose intolerance in mice. METHODS: Mice were divided into four groups: Groups I and II received distilled water and NaAsO2 (10 mg/kg, p.o.) for five weeks, respectively. Groups III and IV were treated with NaAsO2 (10 mg/kg, p.o.) for three weeks, followed by MET (125 and 250 mg/kg, p.o.) for the last two weeks before NaAsO2. A glucose tolerance test was performed on day 35. The serum and tissue parameters were also evaluated. RESULTS: Histopathological examination revealed NaAsO2-induced liver and pancreatic damage. NaAsO2 caused hyperglycemia, glucose intolerance, and a significant increase in liver function enzymes. Administration of NaAsO2 significantly reduced hepatic superoxide dismutase, catalase, glutathione peroxidase, and total thiol levels and increased the content of reactive thiobarbituric acid substances. In addition, it led to an increase in liver nitric oxide levels and protein expression of tumor necrosis factor-α, nuclear factor kappa B, and cysteine-aspartic proteases-3. In contrast, treatment with MET (250 mg/kg) significantly improved NaAsO2-induced biochemical and histopathological changes. CONCLUSION: Our findings suggest that the significant effects of MET against NaAsO2-induced hepatotoxicity and glucose intolerance may be exerted via the regulation of oxidative stress, followed by suppression of inflammation and apoptosis.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Diabetes Mellitus , Intolerância à Glucose , Metformina , Camundongos , Animais , Metformina/farmacologia , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/tratamento farmacológico , Estresse Oxidativo , Apoptose , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle
4.
Diabetes ; 72(11): 1534-1546, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37552863

RESUMO

It is well established that chronic glucocorticoid exposure causes hyperglycemia. While glucocorticoid receptor (GR) stimulates hepatic gluconeogenic gene transcription, additional mechanisms are activated by chronic glucocorticoid exposure to enhance gluconeogenesis. We found that chronic glucocorticoid treatment activated sphingosine-1-phosphate (S1P)-mediated signaling. Hepatic knockdown of hepatic S1P receptor 1 (S1PR1) had no effect on chronic glucocorticoid-induced glucose intolerance but elevated fasting plasma insulin levels. In contrast, hepatic S1PR3 knockdown exacerbated chronic glucocorticoid-induced glucose intolerance without affecting fasting plasma insulin levels. Finally, hepatic S1PR2 knockdown attenuated chronic glucocorticoid-induced glucose intolerance and reduced fasting plasma insulin levels. Here, we focused on dissecting the role of S1PR2 signaling in chronic glucocorticoid response on glucose homeostasis. We found that chronic glucocorticoid-induced hepatic gluconeogenesis, gluconeogenic gene expression, and GR recruitment to the glucocorticoid response elements (GREs) of gluconeogenic genes were all reduced in hepatic S1PR2 knockdown male mice. Hepatic S1PR2 knockdown also enhanced glucocorticoid suppression of RAR-related orphan receptor γ (RORγ) expression. Hepatic RORγ overexpression in hepatic S1PR2 knockdown mice restored glucocorticoid-induced glucose intolerance, gluconeogenic gene expression, and GR recruitment to their GREs. Conversely, RORγ antagonist and the reduction of hepatic RORγ expression attenuated such glucocorticoid effects. Thus, chronic glucocorticoid exposure induces an S1PR2-RORγ axis to cooperate with GR to enhance hepatic gluconeogenesis. Overall, this work provides novel mechanisms of and pharmaceutical targets against steroid-induced hyperglycemia.


Assuntos
Intolerância à Glucose , Hiperglicemia , Insulinas , Hepatopatias , Camundongos , Masculino , Animais , Glucocorticoides/metabolismo , Gluconeogênese/genética , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Fígado/metabolismo , Hiperglicemia/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Insulinas/metabolismo
5.
Part Fibre Toxicol ; 20(1): 25, 2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-37400850

RESUMO

BACKGROUND: We previously found that air pollution particles reaching the gastrointestinal tract elicit gut inflammation as shown by up-regulated gene expression of pro-inflammatory cytokines and monocyte/macrophage markers. This inflammatory response was associated with beta-cell dysfunction and glucose intolerance. So far, it remains unclear whether gut inflammatory changes upon oral air pollution exposure are causally linked to the development of diabetes. Hence, our aim was to assess the role of immune cells in mediating glucose intolerance instigated by orally administered air pollutants. METHODS: To assess immune-mediated mechanisms underlying air pollution-induced glucose intolerance, we administered diesel exhaust particles (DEP; NIST 1650b, 12 µg five days/week) or phosphate-buffered saline (PBS) via gavage for up to 10 months to wild-type mice and mice with genetic or pharmacological depletion of innate or adaptive immune cells. We performed unbiased RNA-sequencing of intestinal macrophages to elucidate signaling pathways that could be pharmacologically targeted and applied an in vitro approach to confirm these pathways. RESULTS: Oral exposure to air pollution particles induced an interferon and inflammatory signature in colon macrophages together with a decrease of CCR2- anti-inflammatory/resident macrophages. Depletion of macrophages, NLRP3 or IL-1ß protected mice from air pollution-induced glucose intolerance. On the contrary, Rag2-/- mice lacking adaptive immune cells developed pronounced gut inflammation and glucose intolerance upon oral DEP exposure. CONCLUSION: In mice, oral exposure to air pollution particles triggers an immune-mediated response in intestinal macrophages that contributes to the development of a diabetes-like phenotype. These findings point towards new pharmacologic targets in diabetes instigated by air pollution particles.


Assuntos
Intolerância à Glucose , Emissões de Veículos , Camundongos , Animais , Emissões de Veículos/toxicidade , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Intolerância à Glucose/induzido quimicamente , Inflamação , Imunidade Inata
6.
Food Chem Toxicol ; 178: 113924, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37393015

RESUMO

Arsenic is a toxic metalloid that increases the risk of hepatotoxicity and hyperglycemia. The objective of the present study was to assess the effect of ferulic acid (FA) in mitigating glucose intolerance and hepatotoxicity caused by sodium arsenite (SA). A total of six groups including control, FA 100 mg/kg, SA 10 mg/kg, and groups that received different doses of FA (10, 30, and 100 mg/kg), respectively just before SA (10 mg/kg) for 28 days were examined. Fasting blood sugar (FBS) and glucose tolerance tests were conducted on the 29th day. On day 30, mice were sacrificed and blood and tissues (liver and pancreas) were collected for further investigations. FA reduced FBS and improved glucose intolerance. Liver function and histopathological studies confirmed that FA preserved the structure of the liver in groups received SA. Furthermore, FA increased antioxidant defense and decreased lipid peroxidation and tumor necrosis factor-alpha level in SA-treated mice. FA, at the doses of 30 and 100 mg/kg, prevented the decrease in the expression of PPAR-γ and GLUT2 proteins in the liver of mice exposed to SA. In conclusion, FA prevented SA-induced glucose intolerance and hepatotoxicity by reducing oxidative stress, inflammation, and hepatic overexpression of PPAR-γ and GLUT2 proteins.


Assuntos
Arsênio , Doença Hepática Induzida por Substâncias e Drogas , Intolerância à Glucose , Camundongos , Animais , Arsênio/toxicidade , Arsênio/metabolismo , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/tratamento farmacológico , Intolerância à Glucose/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Antioxidantes/farmacologia , Fígado , Estresse Oxidativo , Hipoglicemiantes/farmacologia , Hipoglicemiantes/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/metabolismo
8.
Pestic Biochem Physiol ; 192: 105391, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37105618

RESUMO

The use of arsenic in arsenic-based pesticides has been common in many countries in the past and today. There is considerable evidence linking arsenic exposure to hepatotoxicity and diabetes. Destructive phenomena such as hepatic oxidative stress and inflammation can interfere with glucose uptake and insulin function. In the present study, the antioxidant, anti-inflammatory, and molecular mechanism of citicoline against sodium arsenite-induced hepatotoxicity and glucose intolerance were investigated in mice. Citicoline improved glucose tolerance impaired by sodium arsenite. Citicoline increased the hepatic activity of catalase, superoxide dismutase, and glutathione peroxidase enzymes. Moreover, we found that citicoline prevents an increase in the levels of thiobarbituric acid reactive substances. Citicoline reduced levels of caspase 3, tumor necrosis factor-alpha, and interleukin 6 in sodium arsenite intoxicated groups. It was shown that citicoline increased the expression of arsenite methyltransferase, vesicle-associated membrane protein 2, peroxisome proliferator-activated receptor gamma, and sirtuin 3 to combat sodium arsenite toxicity. Citicoline reduced glucose intolerance, which was disrupted by sodium arsenite, by affecting the pancreatic and extra-pancreatic pathways involved in insulin production, secretion, and action. Based on our results, citicoline can be considered a modulating agent against arsenic-induced hepatotoxicity and hyperglycemia. Considering the relationship between arsenic exposure and the occurrence of side effects such as liver toxicity and diabetes, it is necessary to monitor and awareness of arsenic residues from sources such as drinking water.


Assuntos
Arsênio , Doença Hepática Induzida por Substâncias e Drogas , Diabetes Mellitus , Intolerância à Glucose , Insulinas , Sirtuína 3 , Camundongos , Animais , Arsênio/toxicidade , Arsênio/metabolismo , Sirtuína 3/efeitos adversos , Sirtuína 3/metabolismo , Proteína 2 Associada à Membrana da Vesícula/metabolismo , Proteína 2 Associada à Membrana da Vesícula/farmacologia , PPAR gama/metabolismo , Citidina Difosfato Colina/efeitos adversos , Citidina Difosfato Colina/metabolismo , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/tratamento farmacológico , Diabetes Mellitus/induzido quimicamente , Antioxidantes/farmacologia , Estresse Oxidativo , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Insulinas/efeitos adversos , Insulinas/metabolismo , Metiltransferases
9.
Biochem Pharmacol ; 210: 115486, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36893817

RESUMO

BACKGROUND: Exogenous glucocorticoids (CGs) possess relevant therapeutic effects but exert diabetogenic actions when in excess. Thus, ligands with potential therapeutic applications and fewer adverse effects are needed. To this, we analyzed whether mometasone furoate (MF), a CG expected to cause fewer side effects, given through systemic routes, could maintain the anti-inflammatory actions without relevant repercussions on metabolism. METHODS: The anti-inflammatory effect of MF was evaluated with both peritonitis and colitis models in rodents. Glucose and lipid metabolism were investigated in male and female rats treated daily with MF with different doses and routes of administration for seven days. The involvement of glucocorticoid receptor (GR) on MF actions was assessed in animals pretreated with mifepristone. Also, the potential reversibility of the adverse effects was assessed. Dexamethasone was used as a positive control. RESULTS: MF treatment resulted in glucose intolerance in male rats treated through intraperitoneal (ip) but not oral gavage route (og). In female rats, none of the routes led to glucose intolerance. MF treatment attenuated insulin sensitivity and increased pancreatic ß-cell mass, regardless of the sex and route of administration. MF treatment through og route did not result in dyslipidemia, as observed in rats treated through the ip route (both sexes). The anti-inflammatory and metabolic adverse effects of MF were GR-dependent, and metabolic outcomes altered by MF administration were reversible. CONCLUSION: MF maintains anti-inflammatory activity when administered by systemic routes and exerts less impact on metabolism when administered orally in male and female rats, effects that are GR-dependent and reversible. Category: Metabolic Disorders and Endocrinology.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Intolerância à Glucose , Pregnadienodiois , Masculino , Feminino , Ratos , Animais , Furoato de Mometasona , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/tratamento farmacológico , Pregnadienodiois/efeitos adversos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Glucocorticoides/toxicidade , Administração por Inalação
10.
Sci Total Environ ; 877: 162934, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-36934930

RESUMO

Results of human and animal studies independently suggest that either ambient fine particulate matter (PM2.5) air pollution exposure or a disturbed circadian rhythm (circadian dyssynchrony) are important contributing factors to the rapidly evolving type-2-diabetes (T2D) epidemic. The objective of this study is to investigate whether circadian dyssynchrony increases the susceptibility to PM2.5 and how PM2.5 affects metabolic health in circadian dyssynchrony. We examined systemic and organ-specific changes in glucose homeostasis and insulin sensitivity in mice maintained on a regular (12/12 h light/dark) or disrupted (18/6 h light/dark, light-induced circadian dyssynchrony, LICD) light cycle exposed to air or concentrated PM2.5 (CAP, 6 h/day, 30 days). Exposures during Zeitgeber ZT3-9 or ZT11-17 (Zeitgeber in circadian time, ZT0 = begin of light cycle) tested for time-of-day PM2.5 sensitivity (chronotoxicity). Mice transgenic for lung-specific overexpression of extracellular superoxide dismutase (ecSOD-Tg) were used to assess the contribution of CAP-induced pulmonary oxidative stress. Both, CAP exposure from ZT3-9 or ZT11-17, decreased glucose tolerance and insulin sensitivity in male mice with LICD, but not in female mice or in mice kept on a regular light cycle. Although changes in glucose homeostasis in CAP-exposed male mice with LICD were not associated with obesity, they were accompanied by white adipose tissue (WAT) inflammation, impaired insulin signaling in skeletal muscle and liver, and systemic and pulmonary oxidative stress. Preventing CAP-induced oxidative stress in the lungs mitigated the CAP-induced decrease in glucose tolerance and insulin sensitivity in LICD. Our results demonstrate that circadian dyssynchrony is a novel susceptibility state for PM2.5 and suggest that PM2.5 by inducing pulmonary oxidative stress increases glucose intolerance and insulin resistance in circadian dyssynchrony.


Assuntos
Poluentes Atmosféricos , Intolerância à Glucose , Resistência à Insulina , Humanos , Masculino , Feminino , Camundongos , Animais , Material Particulado/toxicidade , Material Particulado/metabolismo , Intolerância à Glucose/induzido quimicamente , Pulmão , Estresse Oxidativo , Glucose/metabolismo , Poluentes Atmosféricos/toxicidade , Poluentes Atmosféricos/metabolismo
11.
Planta Med ; 88(9-10): 735-744, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35777366

RESUMO

Diabetes is a life-threatening and debilitating disease with pathological hallmarks, including glucose intolerance and insulin resistance. Plant compounds are a source of novel and effective therapeutics, and the flavonoid (-)-epicatechin, common to popular foods worldwide, has been shown to improve carbohydrate metabolism in both clinical studies and preclinical models. We hypothesized that (-)-epicatechin would alleviate thermoneutral housing-induced glucose intolerance. Male rats were housed at either thermoneutral (30 °C) or room temperature (24 °C) for 16 weeks and gavaged with either 1 mg/kg body weight or vehicle for the last 15 days before sacrifice. Rats housed at thermoneutrality had a significantly elevated serum glucose area under the curve (p < 0.05) and reduced glucose-mediated insulin secretion. In contrast, rats at thermoneutrality treated with (-)-epicatechin had improved glucose tolerance and increased insulin secretion (p < 0.05). Insulin tolerance tests revealed no differences in insulin sensitivity in any of the four groups. Pancreatic immunohistochemistry staining showed significantly greater islet insulin positive cells in animals housed at thermoneutrality. In conclusion, (-)-epicatechin improved carbohydrate tolerance via increased insulin secretion in response to glucose challenge without a change in insulin sensitivity.


Assuntos
Catequina , Intolerância à Glucose , Resistência à Insulina , Animais , Glicemia/metabolismo , Catequina/farmacologia , Glucose/farmacologia , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/tratamento farmacológico , Habitação , Insulina , Resistência à Insulina/fisiologia , Ratos
12.
Life Sci ; 305: 120770, 2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-35792179

RESUMO

AIMS: Epidemic obesity and diabetes have led to increased use of low-calorie sweeteners. Although several studies have suggested that consumption of artificial sweeteners, such as aspartame and saccharin, might have negative effects, the potential impacts of natural sweeteners on human health remain largely unknown. MAIN METHODS: The deferential effects of short term and long term consumption of sorbitol on glucose homeostasis in mice by oral gavage. The glucose homeostasis and utility were evaluated by both oral or intraperitoneal glucose tolerance tests. Insulin levels were determined using enzyme-linked immunosorbent assay. Changes of gut microbiome were evaluated by 16S rRNA gene sequencing, and analyzed by principal components analysis. KEY FINDINGS: Bolus feeding of sorbitol by gavage significantly increased plasma insulin concentrations and decreased fasting blood glucose levels. Intriguingly, long-term sorbitol gavage for four weeks showed no significant effects on intraperitoneal glucose tolerance test outcomes, but it induced glucose intolerance according to the oral glucose tolerance test. Thus, we tested whether long-term sorbitol gavage might alter the relative abundances of gut microbiome constituents in mice. Principal components analysis indicated that long-term sorbitol intake indeed caused significant changes to the gut microbiome. In particular, we found that long-term sorbitol intake significantly decreased the relative abundances of Bifidobacterium, Lachnospiraceae UCG 001, Lachnospiraceae NK4A136, Eubacterium ventriosum, Candidatus Arthromitus, and Ruminococcus torques. We also found that long-term sorbitol increased the relative abundances of Helicobacter, Tyzzerella, Alistipes, and Prevotella 9. SIGNIFICANCE: Long-term sorbitol consumption may change the composition of the gut microbiome and potentially induce glucose intolerance.


Assuntos
Microbioma Gastrointestinal , Intolerância à Glucose , Insulinas , Animais , Glicemia , Glucose/farmacologia , Intolerância à Glucose/induzido quimicamente , Humanos , Insulinas/farmacologia , Camundongos , RNA Ribossômico 16S/genética , Sorbitol/farmacologia , Edulcorantes/efeitos adversos
14.
Int Urol Nephrol ; 54(10): 2555-2566, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35277831

RESUMO

OBJECTIVE: To explore the efficacy and safety of tacrolimus (TAC) combined with corticosteroids in patients with idiopathic membranous nephropathy (IMN). METHODS: A literature search was performed using Embase, Cochrane Library and PubMed from inception through May 31, 2021. All randomized controlled trials (RCTs) exploring the efficacy and safety of TAC combined with corticosteroids in IMN patients were included based on the inclusion and exclusion criteria. Data analyses were conducted using RevMan software (version 5.4). RESULTS: Seven RCTs involving 520 patients were included in this meta-analysis. Compared with control treatment, TAC combined with corticosteroids could significantly increase the complete remission (CR) rate, total remission (TR) rate, and serum albumin levels, as well as decrease the proteinuria levels within 6-month treatment, but the advantage did not persist to 12-month treatment. After 18-month treatment, the effect of TAC combined with corticosteroids on increasing CR rate, TR rate, and serum albumin levels was significantly worse than control treatment. The mean time to remission in TAC combined corticosteroids group was significantly shorter than that in the control group. The relapse rate, no response rate, change in estimate of the glomerular filtration rate (eGFR), and overall incidence of adverse reactions showed no significant difference between TAC combined with corticosteroids group and control group. However, TAC combined with corticosteroids did have a higher risk of hand tremor, nephrotoxicity, and glucose intolerance than control treatment. CONCLUSION: TAC combined with corticosteroids has a significant therapeutic effect for IMN patients within 1-year treatment, especially in the first 6 months. However, in the longer-term treatment, TAC combined with corticosteroids does not have an advantage. TAC combined with corticosteroids has a higher risk of hand tremor, nephrotoxicity, and glucose intolerance. More high-quality studies are needed to further verify the long-term efficacy and safety of TAC combined with glucocorticoids in IMN patients.


Assuntos
Corticosteroides , Glomerulonefrite Membranosa , Tacrolimo , Corticosteroides/efeitos adversos , Corticosteroides/uso terapêutico , Quimioterapia Combinada , Glomerulonefrite Membranosa/complicações , Glomerulonefrite Membranosa/tratamento farmacológico , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/tratamento farmacológico , Humanos , Imunossupressores/efeitos adversos , Imunossupressores/uso terapêutico , Ensaios Clínicos Controlados Aleatórios como Assunto , Albumina Sérica/análise , Tacrolimo/efeitos adversos , Tacrolimo/uso terapêutico , Resultado do Tratamento , Tremor/induzido quimicamente , Tremor/tratamento farmacológico
15.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35145023

RESUMO

Insulin resistance and ß-cell dysfunction are two main molecular bases yet to be further elucidated for type 2 diabetes (T2D). Accumulating evidence indicates that stimulator of interferon genes (STING) plays an important role in regulating insulin sensitivity. However, its function in ß-cells remains unknown. Herein, using global STING knockout (STING-/-) and ß-cell-specific STING knockout (STING-ßKO) mouse models, we revealed a distinct role of STING in the regulation of glucose homeostasis through peripheral tissues and ß-cells. Specially, although STING-/- beneficially alleviated insulin resistance and glucose intolerance induced by high-fat diet, it surprisingly impaired islet glucose-stimulated insulin secretion (GSIS). Importantly, STING is decreased in islets of db/db mice and patients with T2D, suggesting a possible role of STING in ß-cell dysfunction. Indeed, STING-ßKO caused glucose intolerance due to impaired GSIS, indicating that STING is required for normal ß-cell function. Islet transcriptome analysis showed that STING deficiency decreased expression of ß-cell function-related genes, including Glut2, Kcnj11, and Abcc8, contributing to impaired GSIS. Mechanistically, the assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and cleavage under targets and tagmentation (CUT&Tag) analyses suggested that Pax6 was the transcription factor that might be associated with defective GSIS in STING-ßKO mice. Indeed, Pax6 messenger RNA and protein levels were down-regulated and its nuclear localization was lost in STING-ßKO ß-cells. Together, these data revealed a function of STING in the regulation of insulin secretion and established pathophysiological significance of fine-tuned STING within ß-cells and insulin target tissues for maintaining glucose homeostasis.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Intolerância à Glucose/induzido quimicamente , Glucose/metabolismo , Insulina/metabolismo , Proteínas de Membrana/metabolismo , Animais , Diabetes Mellitus Experimental , Dieta Hiperlipídica/efeitos adversos , Regulação para Baixo , Regulação da Expressão Gênica , Homeostase , Humanos , Insulina/sangue , Resistência à Insulina , Células Secretoras de Insulina , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout
16.
Sci Total Environ ; 826: 154095, 2022 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-35219660

RESUMO

The prevalence of diabetes in children and adolescents has been rising gradually, which is relevant to adverse environment during development, especially prepartum. We aimed to explore the effects of prenatal dexamethasone exposure (PDE) on ß-cell function and glucose homeostasis in juvenile offspring rats. Pregnant Wistar rats were subcutaneously administered with dexamethasone [0.1, 0.2, 0.4mg/(kg.d)] from gestational day 9 to 20. PDE impaired glucose tolerance in the male offspring rather than the females. In male offspring, PDE impaired the development and function of ß-cells, accompanied with lower H3K9ac, H3K14ac and H3K27ac levels in the promoter region of angiotensin-converting enzyme 2 (ACE2) as well as suppressed ACE2 expression. Meanwhile, PDE increased expression of glucocorticoid receptor (GR) and histone deacetylase 3 (HDAC3) in fetal pancreas. Dexamethasone also inhibited ACE2 expression and insulin production in vitro. Recombinant expression of ACE2 restored insulin production inhibited by dexamethasone. In addition, dexamethasone activated GR and HDAC3, increased protein interaction of GR with HDAC3, and promoted the binding of GR-HDAC3 complex to ACE2 promoter region. Both RU486 and TSA abolished dexamethasone-induced decline of histone acetylation and ACE2 expression. In summary, suppression of ACE2 is involved in PDE induced ß-cell dysfunction and glucose intolerance in juvenile male offspring rats.


Assuntos
Intolerância à Glucose , Insulinas , Efeitos Tardios da Exposição Pré-Natal , Adolescente , Enzima de Conversão de Angiotensina 2 , Animais , Dexametasona/toxicidade , Repressão Epigenética , Feminino , Intolerância à Glucose/induzido quimicamente , Humanos , Insulinas/metabolismo , Insulinas/toxicidade , Masculino , Pâncreas/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Ratos , Ratos Wistar , Receptores de Glucocorticoides
17.
Int J Mol Sci ; 22(24)2021 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-34948019

RESUMO

The NAD-dependent deacetylase SIRT1 improves ß cell function. Accordingly, nicotinamide mononucleotide (NMN), the product of the rate-limiting step in NAD synthesis, prevents ß cell dysfunction and glucose intolerance in mice fed a high-fat diet. The current study was performed to assess the effects of NMN on ß cell dysfunction and glucose intolerance that are caused specifically by increased circulating free fatty acids (FFAs). NMN was intravenously infused, with or without oleate, in C57BL/6J mice over a 48-h-period to elevate intracellular NAD levels and consequently increase SIRT1 activity. Administration of NMN in the context of elevated plasma FFA levels considerably improved glucose tolerance. This was due not only to partial protection from FFA-induced ß cell dysfunction but also, unexpectedly, to a significant decrease in insulin clearance. However, in conditions of normal FFA levels, NMN impaired glucose tolerance due to decreased ß cell function. The presence of this dual action of NMN suggests caution in its proposed therapeutic use in humans.


Assuntos
Ácidos Graxos não Esterificados/sangue , Intolerância à Glucose/tratamento farmacológico , Glucose/efeitos adversos , Insulina/metabolismo , Mononucleotídeo de Nicotinamida/administração & dosagem , Ácido Oleico/efeitos adversos , Animais , Intolerância à Glucose/sangue , Intolerância à Glucose/induzido quimicamente , Células Hep G2 , Humanos , Infusões Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NAD/metabolismo , Mononucleotídeo de Nicotinamida/farmacologia , Sirtuína 1/metabolismo , Regulação para Cima
18.
Environ Sci Pollut Res Int ; 28(44): 62105-62115, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34590231

RESUMO

In the present work, a systematic review and meta-analysis were performed to examine the probable relation between maternal exposure to bisphenol A (BPA), as estrogen-disrupting compounds, and gestational diabetes mellitus (GDM), and impaired glucose tolerance (IGT). We comprehensively searched three electronic databases to retrieve published studies on maternal exposure to BPA and GDM/IGT, through February 2021. Cochran's Q test and I2 statistics were employed for testing heterogeneity across studies. DerSimonian and Liard random-effects model was used to determine the pooled estimates. Otherwise, the fixed-effects model with inverse-variance weights was applied. Sensitivity analysis was performed to determine the robustness of the results by excluding each study from the pooled estimate. The potential publication bias was examined using Begg's and Egger's tests. The pooled odds ratio did not show BPA exposure to be a significant risk factor for GDM (OR = 0.90, 95% CI = 0.62-1.33, I2: 50.7%). Also, no significant association was observed between BPA exposure and risk of IGT (OR = 0.93, 95% CI = 0.40-2.18, I2: 11.5%). Based on the findings of this study, no association was found between exposure to BPA during pregnancy and the risk of GDM/IGT. Albeit no heterogeneity was found between studies.


Assuntos
Intolerância à Glucose , Compostos Benzidrílicos , Feminino , Glucose , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/epidemiologia , Humanos , Fenóis/toxicidade , Gravidez
19.
Cancer Chemother Pharmacol ; 88(5): 857-865, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34350479

RESUMO

PURPOSE: Cytotoxic chemotherapy-induced lung injury is a fatal complication in patients with lung cancer and interstitial lung disease (ILD). We aimed to evaluate the association between hyperglycemia and this form of lung injury in patients with lung cancer concomitant with ILD. METHODS: From 1147 patients with advanced lung cancer, we retrospectively enrolled 98 patients with ILD whose hemoglobin A1c (HbA1c) levels were measured, and investigated the association between HbA1c levels and cytotoxic chemotherapy-induced lung injury. In 73 patients whose serum samples were retained, we measured serum levels of advanced glycation end products (AGE) and assessed the association of AGE levels with HbA1c levels and cytotoxic chemotherapy-induced lung injury. RESULTS: The incidence of cytotoxic chemotherapy-induced lung injury was significantly higher in patients with HbA1c levels ≥ 5.8% than in those with HbA1c levels < 5.8%, but not in those with HbA1c levels ≥ 6.5% than in those with HbA1c levels < 6.5%. The multivariate logistic regression model revealed that HbA1c level ≥ 5.8% was a significant risk factor for this complication [odds ratio 3.178 (95% confidence interval 1.057-9.556), P = 0.040]. In addition, serum AGE levels were significantly higher in patients with HbA1c levels ≥ 5.8% than in those with HbA1c levels < 5.8% [median (interquartile range); 0.129 (0.023-0.290) and 0.474 (0.213-1.109) µg/mL, P = 0.001]. CONCLUSION: Glucose intolerance (e.g., HbA1c level ≥ 5.8%) may be a risk factor of cytotoxic chemotherapy-induced lung injury, which might be associated with elevated AGE production due to hyperglycemia.


Assuntos
Antineoplásicos/efeitos adversos , Intolerância à Glucose/induzido quimicamente , Doenças Pulmonares Intersticiais/tratamento farmacológico , Lesão Pulmonar/induzido quimicamente , Neoplasias Pulmonares/tratamento farmacológico , Idoso , Feminino , Hemoglobinas Glicadas/análise , Produtos Finais de Glicação Avançada/sangue , Humanos , Hiperglicemia/induzido quimicamente , Hiperglicemia/fisiopatologia , Doenças Pulmonares Intersticiais/complicações , Doenças Pulmonares Intersticiais/fisiopatologia , Neoplasias Pulmonares/fisiopatologia , Masculino , Estudos Retrospectivos , Capacidade Vital
20.
Int J Mol Sci ; 22(13)2021 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-34281282

RESUMO

Urinary acrolein adduct levels have been reported to be increased in both habitual smokers and type-2 diabetic patients. The impairment of glucose transport in skeletal muscles is a major factor responsible for glucose uptake reduction in type-2 diabetic patients. The effect of acrolein on glucose metabolism in skeletal muscle remains unclear. Here, we investigated whether acrolein affects muscular glucose metabolism in vitro and glucose tolerance in vivo. Exposure of mice to acrolein (2.5 and 5 mg/kg/day) for 4 weeks substantially increased fasting blood glucose and impaired glucose tolerance. The glucose transporter-4 (GLUT4) protein expression was significantly decreased in soleus muscles of acrolein-treated mice. The glucose uptake was significantly decreased in differentiated C2C12 myotubes treated with a non-cytotoxic dose of acrolein (1 µM) for 24 and 72 h. Acrolein (0.5-2 µM) also significantly decreased the GLUT4 expression in myotubes. Acrolein suppressed the phosphorylation of glucose metabolic signals IRS1, Akt, mTOR, p70S6K, and GSK3α/ß. Over-expression of constitutive activation of Akt reversed the inhibitory effects of acrolein on GLUT4 protein expression and glucose uptake in myotubes. These results suggest that acrolein at doses relevant to human exposure dysregulates glucose metabolism in skeletal muscle cells and impairs glucose tolerance in mice.


Assuntos
Acroleína/toxicidade , Transportador de Glucose Tipo 4/antagonistas & inibidores , Glucose/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Acroleína/administração & dosagem , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Glicemia/metabolismo , Linhagem Celular , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Humanos , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...